Skip to main content

Advertisement

Log in

Biomarkers for Risk Prediction in Acute Decompensated Heart Failure

  • Decompensated Heart Failure (JE Ho, Section Editor)
  • Published:
Current Heart Failure Reports Aims and scope Submit manuscript

Abstract

Risk prediction in patients admitted with acute decompensated heart failure (ADHF) remains a challenge. Biomarkers may improve risk prediction, which in turn may help to better inform patients regarding short-term and long-term prognosis, therapy and care. Most data on biomarkers have been derived from patient cohorts with chronic heart failure. In ADHF, currently, risk tools largely rely on common clinical and biochemical parameters. However, ADHF is not a single disease. It presents in various manners and different etiologies may underlie ADHF, which are reflected by different biomarkers. In the last decade, many studies have reported the prognostic value of these biomarkers. These studies have attempted to describe a value for statistical modeling, e.g., reclassification indices, in an effort to report incremental value over a clinical model or the “gold standard”. However, the overall incremental predictive value of biomarkers has been modest compared to already existing clinical models. Natriuretic peptides, e.g., (NTpro-)BNP, are the benchmark, but head-to-head comparisons show that there are novel biomarkers with comparable prognostic value. Multimarker strategies may provide superior risk stratification. Future studies should elucidate cost-effectiveness of single or combined biomarker testing. The purpose of this review was to provide an update on current biomarkers and to identify new promising biomarkers than can be used in prognostication of acute heart failure.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1

Similar content being viewed by others

References

Papers of particular interest, published recently, have been highlighted as: • Of importance •• Of major importance

  1. Follath F, Yilmaz MB, Delgado JF, et al. Clinical presentation, management and outcomes in the Acute Heart Failure Global Survey of Standard Treatment (ALARM-HF). Intensive Care Med. 2011;37:619–26. ALARM-HF was a large survey among 4,953 patients with AHF in nine different countries and described characteristics and management of acute heart failure in these patients.

    CAS  PubMed  Google Scholar 

  2. Rudiger A, Harjola VP, Muller A, et al. Acute heart failure: clinical presentation, one-year mortality and prognostic factors. Eur J Heart Fail. 2005;7:662–70.

    PubMed  Google Scholar 

  3. Harjola VP, Follath F, Nieminen MS, et al. Characteristics, outcomes, and predictors of mortality at 3 months and 1 year in patients hospitalized for acute heart failure. Eur J Heart Fail. 2010;12:239–48. The EuroHeart Failure Survey (EHFS) II evaluated 3-month and 12-month mortality in 2,981 patients with acute heart failure from 30 different countries and identified clinical risk markers.

    PubMed  Google Scholar 

  4. Biomarkers Definitions Working Group. Biomarkers and surrogate endpoints: preferred definitions and conceptual framework. Clin Pharmacol Ther. 2001;69:89–95. The Biomarkers Definitions Working Group defined biomarkers as a characteristic that is objectively measured and evaluated as an indicator of normal biological processes, pathogenic processes or pharmacologic responses to a therapeutic intervention.

    Google Scholar 

  5. Lee CS, Tkacs NC. Current concepts of neurohormonal activation in heart failure: mediators and mechanisms. AACN Adv Crit Care. 2008;19:364–85. quiz 386-7.

    PubMed  Google Scholar 

  6. Misra A, Mann DL. Treatment of heart failure beyond practice guidelines. Role of cardiac remodeling. Circ J. 2008;72(Suppl A):A1–7.

    PubMed  Google Scholar 

  7. Jougasaki M, Burnett Jr JC. Adrenomedullin: potential in physiology and pathophysiology. Life Sci. 2000;66:855–72.

    CAS  PubMed  Google Scholar 

  8. Sata M, Kakoki M, Nagata D, et al. Adrenomedullin and nitric oxide inhibit human endothelial cell apoptosis via a cyclic GMP-independent mechanism. Hypertension. 2000;36:83–8.

    CAS  PubMed  Google Scholar 

  9. Lainchbury JG, Troughton RW, Lewis LK, Yandle TG, Richards AM, Nicholls MG. Hemodynamic, hormonal, and renal effects of short-term adrenomedullin infusion in healthy volunteers. J Clin Endocrinol Metab. 2000;85:1016–20. This article describes the hemodynamic effects of systemic infusion of adrenomedullin up to pathophysiological plasma levels in healthy volunteers.

    CAS  PubMed  Google Scholar 

  10. Nicholls MG, Charles CJ, Lainchbury JG, et al. Adrenomedullin in heart failure. Hypertens Res. 2003;26(Suppl):S135–40.

    CAS  PubMed  Google Scholar 

  11. Meeran K, O’Shea D, Upton PD, et al. Circulating adrenomedullin does not regulate systemic blood pressure but increases plasma prolactin after intravenous infusion in humans: a pharmacokinetic study. J Clin Endocrinol Metab. 1997;82:95–100.

    CAS  PubMed  Google Scholar 

  12. Morgenthaler NG, Struck J, Alonso C, Bergmann A. Measurement of midregional proadrenomedullin in plasma with an immunoluminometric assay. Clin Chem. 2005;51:1823–9.

    CAS  PubMed  Google Scholar 

  13. Travaglino F, Russo V, De Berardinis B, et al. Thirty and ninety days mortality predictive value of admission and in-hospital procalcitonin and mid-regional pro-adrenomedullin testing in patients with dyspnea. Results from the VERyfing DYspnea trial. Am J Emerg Med. 2014;32:334–41.

    PubMed  Google Scholar 

  14. Cinar O, Cevik E, Acar A, et al. Evaluation of mid-regional pro-atrial natriuretic peptide, procalcitonin, and mid-regional pro-adrenomedullin for the diagnosis and risk stratification of dyspneic ED patients. Am J Emerg Med. 2012;30:1915–20.

    PubMed  Google Scholar 

  15. Shah RV, Truong QA, Gaggin HK, Pfannkuche J, Hartmann O, Januzzi Jr JL. Mid-regional pro-atrial natriuretic peptide and pro-adrenomedullin testing for the diagnostic and prognostic evaluation of patients with acute dyspnoea. Eur Heart J. 2012;33:2197–205.

    CAS  PubMed Central  PubMed  Google Scholar 

  16. Maisel A, Mueller C, Nowak RM, et al. Midregion prohormone adrenomedullin and prognosis in patients presenting with acute dyspnea: results from the BACH (Biomarkers in Acute Heart Failure) trial. J Am Coll Cardiol. 2011;58:1057–67. The BACH trial was a prospective, multicenter, international study of 1,641 ADHF patients presenting with dyspnea at the emergency department and studied the diagnostic and prognostic value of various biomarkers.

    CAS  PubMed  Google Scholar 

  17. Peacock WF, Nowak R, Christenson R, et al. Short-term mortality risk in emergency department acute heart failure. Acad Emerg Med. 2011;18:947–58.

    PubMed  Google Scholar 

  18. Chatterjee K. Neurohormonal activation in congestive heart failure and the role of vasopressin. Am J Cardiol. 2005;95:8B–13. This review focuses on the different components of neurohormonal activation in heart failure.

    CAS  PubMed  Google Scholar 

  19. Francis GS, Benedict C, Johnstone DE, et al. Comparison of neuroendocrine activation in patients with left ventricular dysfunction with and without congestive heart failure. A substudy of the Studies of Left Ventricular Dysfunction (SOLVD). Circulation. 1990;82:1724–9.

    CAS  PubMed  Google Scholar 

  20. Stoiser B, Mortl D, Hulsmann M, et al. Copeptin, a fragment of the vasopressin precursor, as a novel predictor of outcome in heart failure. Eur J Clin Invest. 2006;36:771–8.

    CAS  PubMed  Google Scholar 

  21. Morgenthaler NG, Struck J, Alonso C, Bergmann A. Assay for the measurement of copeptin, a stable peptide derived from the precursor of vasopressin. Clin Chem. 2006;52:112–9.

    CAS  PubMed  Google Scholar 

  22. Struck J, Morgenthaler NG, Bergmann A. Copeptin, a stable peptide derived from the vasopressin precursor, is elevated in serum of sepsis patients. Peptides. 2005;26:2500–4.

    CAS  PubMed  Google Scholar 

  23. Meijer E, Bakker SJ, Halbesma N, de Jong PE, Struck J, Gansevoort RT. Copeptin, a surrogate marker of vasopressin, is associated with microalbuminuria in a large population cohort. Kidney Int. 2010;77:29–36.

    CAS  PubMed  Google Scholar 

  24. Gegenhuber A, Struck J, Dieplinger B, et al. Comparative evaluation of B-type natriuretic peptide, mid-regional pro-A-type natriuretic peptide, mid-regional pro-adrenomedullin, and Copeptin to predict 1-year mortality in patients with acute destabilized heart failure. J Card Fail. 2007;13:42–9. This study compares the prognostic value of different novel biomarkers and concludes some have similar predictive properties compared with BNP for 1-year all-cause mortality.

    CAS  PubMed  Google Scholar 

  25. Neuhold S, Huelsmann M, Strunk G, et al. Prognostic value of emerging neurohormones in chronic heart failure during optimization of heart failure-specific therapy. Clin Chem. 2010;56:121–6.

    CAS  PubMed  Google Scholar 

  26. Miller WL, Hartman KA, Grill DE, Struck J, Bergmann A, Jaffe AS. Serial measurements of midregion proANP and copeptin in ambulatory patients with heart failure: incremental prognostic value of novel biomarkers in heart failure. Heart. 2012;98:389–94. The authors studied serial monitoring of MR-proANP and copeptin and concluded this was useful for detecting the highest-risk outpatients with HF.

    CAS  PubMed Central  PubMed  Google Scholar 

  27. Taupenot L, Harper KL, O’Connor DT. The chromogranin-secretogranin family. N Engl J Med. 2003;348:1134–49. This is an extensive review about the functions and mechanisms of the chromogranin-secretogranin family.

    CAS  PubMed  Google Scholar 

  28. Mazza R, Imbrogno S, Tota B. The interplay between chromogranin A-derived peptides and cardiac natriuretic peptides in cardioprotection against catecholamine-evoked stress. Regul Pept. 2010;165:86–94.

    CAS  PubMed  Google Scholar 

  29. Pieroni M, Corti A, Tota B, et al. Myocardial production of chromogranin A in human heart: a new regulatory peptide of cardiac function. Eur Heart J. 2007;28:1117–27.

    CAS  PubMed  Google Scholar 

  30. Dieplinger B, Gegenhuber A, Struck J, et al. Chromogranin A and C-terminal endothelin-1 precursor fragment add independent prognostic information to amino-terminal proBNP in patients with acute destabilized heart failure. Clin Chim Acta. 2009;400:91–6. In 137 patients with acute destabilized heart failure, increased chromogranin A and CT-proET-1 plasma concentrations add independent prognostic information to NT-proBNP.

    CAS  PubMed  Google Scholar 

  31. Teiger E, Than VD, Richard L, et al. Apoptosis in pressure overload-induced heart hypertrophy in the rat. J Clin Invest. 1996;97:2891–7.

    CAS  PubMed Central  PubMed  Google Scholar 

  32. Kong P, Christia P, Frangogiannis NG. The pathogenesis of cardiac fibrosis. Cell Mol Life Sci. 2014;71:549–74. This review discusses the molecular pathways that are involved in myocardial fibrosis.

    CAS  PubMed  Google Scholar 

  33. Weinberg EO, Shimpo M, De Keulenaer GW, et al. Expression and regulation of ST2, an interleukin-1 receptor family member, in cardiomyocytes and myocardial infarction. Circulation. 2002;106:2961–6.

    CAS  PubMed Central  PubMed  Google Scholar 

  34. Shimpo M, Morrow DA, Weinberg EO, et al. Serum levels of the interleukin-1 receptor family member ST2 predict mortality and clinical outcome in acute myocardial infarction. Circulation. 2004;109:2186–90.

    CAS  PubMed  Google Scholar 

  35. Adams Jr KF, Fonarow GC, Emerman CL, et al. Characteristics and outcomes of patients hospitalized for heart failure in the United States: rationale, design, and preliminary observations from the first 100,000 cases in the Acute Decompensated Heart Failure National Registry (ADHERE). Am Heart J. 2005;149:209–16. The ADHERE registry describes characteristics and outcome of 100,000 patients hospitalized for acute decompensated heart failure.

    PubMed  Google Scholar 

  36. Tago K, Noda T, Hayakawa M, et al. Tissue distribution and subcellular localization of a variant form of the human ST2 gene product, ST2V. Biochem Biophys Res Commun. 2001;285:1377–83.

    CAS  PubMed  Google Scholar 

  37. Sanada S, Hakuno D, Higgins LJ, Schreiter ER, McKenzie AN, Lee RT. IL-33 and ST2 comprise a critical biomechanically induced and cardioprotective signaling system. J Clin Invest. 2007;117:1538–49.

    CAS  PubMed Central  PubMed  Google Scholar 

  38. Seki K, Sanada S, Kudinova AY, et al. Interleukin-33 prevents apoptosis and improves survival after experimental myocardial infarction through ST2 signaling. Circ Heart Fail. 2009;2:684–91.

    CAS  PubMed  Google Scholar 

  39. Januzzi Jr JL, Peacock WF, Maisel AS, et al. Measurement of the interleukin family member ST2 in patients with acute dyspnea: results from the PRIDE (Pro-Brain Natriuretic Peptide Investigation of Dyspnea in the Emergency Department) study. J Am Coll Cardiol. 2007;50:607–13.

    CAS  PubMed  Google Scholar 

  40. Mueller T, Dieplinger B, Gegenhuber A, Poelz W, Pacher R, Haltmayer M. Increased plasma concentrations of soluble ST2 are predictive for 1-year mortality in patients with acute destabilized heart failure. Clin Chem. 2008;54:752–6.

    CAS  PubMed  Google Scholar 

  41. Pascual-Figal DA, Manzano-Fernandez S, Boronat M, et al. Soluble ST2, high-sensitivity troponin T- and N-terminal pro-B-type natriuretic peptide: complementary role for risk stratification in acutely decompensated heart failure. Eur J Heart Fail. 2011;13:718–25. This study investigated the use of ST2 and hs-Troponin and concluded these markers provide superior risk stratification over NT-proBNP.

    CAS  PubMed  Google Scholar 

  42. Shah RV, Chen-Tournoux AA, Picard MH, van Kimmenade RR, Januzzi JL. Serum levels of the interleukin-1 receptor family member ST2, cardiac structure and function, and long-term mortality in patients with acute dyspnea. Circ Heart Fail. 2009;2:311–9.

    CAS  PubMed  Google Scholar 

  43. Shah RV, Januzzi Jr JL. ST2: a novel remodeling biomarker in acute and chronic heart failure. Curr Heart Fail Rep. 2010;7:9–14.

    CAS  PubMed  Google Scholar 

  44. Breidthardt T, Balmelli C, Twerenbold R, et al. Heart failure therapy-induced early ST2 changes may offer long-term therapy guidance. J Card Fail. 2013;19:821–8.

    CAS  PubMed  Google Scholar 

  45. Gaggin HK, Szymonifka J, Bhardwaj A, et al. Head-to-head comparison of serial soluble ST2, growth differentiation factor-15, and highly-sensitive troponin T measurements in patients with chronic heart failure. JACC Heart Fail. 2014;2:65–72. This article performed a head-to-head comparison of three promising biomarkers in chronic heart failure and concluded only serial ST2 measurements added prognostic information to baseline values.

    PubMed  Google Scholar 

  46. Sharma UC, Pokharel S, van Brakel TJ, et al. Galectin-3 marks activated macrophages in failure-prone hypertrophied hearts and contributes to cardiac dysfunction. Circulation. 2004;110:3121–8.

    CAS  PubMed  Google Scholar 

  47. de Boer RA, Voors AA, Muntendam P, van Gilst WH, van Veldhuisen DJ. Galectin-3: a novel mediator of heart failure development and progression. Eur J Heart Fail. 2009;11:811–7. This review describes the mechanisms of galectin-3 in development of heart failure.

    PubMed  Google Scholar 

  48. Calvier L, Miana M, Reboul P, et al. Galectin-3 mediates aldosterone-induced vascular fibrosis. Arterioscler Thromb Vasc Biol. 2013;33:67–75. The authors conclude that aldosterone might induce fibrosis via a galectin-3 dependent pathway.

    CAS  PubMed  Google Scholar 

  49. Sasaki T, Brakebusch C, Engel J, Timpl R. Mac-2 binding protein is a cell-adhesive protein of the extracellular matrix which self-assembles into ring-like structures and binds beta1 integrins, collagens and fibronectin. EMBO J. 1998;17:1606–13.

    CAS  PubMed Central  PubMed  Google Scholar 

  50. Song L, Tang JW, Owusu L, Sun MZ, Wu J, Zhang J. Galectin-3 in cancer. Clin Chim Acta. 2014;431C:185–91.

    Google Scholar 

  51. Henderson NC, Sethi T. The regulation of inflammation by galectin-3. Immunol Rev. 2009;230:160–71.

    CAS  PubMed  Google Scholar 

  52. Carrasco-Sanchez FJ, Aramburu-Bodas O, Salamanca-Bautista P, et al. Predictive value of serum galectin-3 levels in patients with acute heart failure with preserved ejection fraction. Int J Cardiol. 2013;169:177–82.

    PubMed  Google Scholar 

  53. Meijers WC, Januzzi JL, de Filippi C, et al. Elevated plasma galectin-3 is associated with near-term rehospitalization in heart failure: a pooled analysis of 3 clinical trials. Am Heart J. 2014;167:853–60.

  54. van Kimmenade RR, Januzzi Jr JL, Ellinor PT, et al. Utility of amino-terminal pro-brain natriuretic peptide, galectin-3, and apelin for the evaluation of patients with acute heart failure. J Am Coll Cardiol. 2006;48:1217–24. This substudy of the PRIDE demonstrated the clinical utility of galectin-3 for the first time, showing that galectin-3 was the best predictor for 60-day mortality.

    PubMed  Google Scholar 

  55. Eurlings LW, Sanders-van Wijk S, van Kimmenade R, et al. Multimarker strategy for short-term risk assessment in patients with dyspnea in the emergency department: the MARKED (Multi mARKer Emergency Dyspnea)-risk score. J Am Coll Cardiol. 2012;60:1668–77.

    PubMed  Google Scholar 

  56. van der Velde AR, Gullestad L, Ueland T, et al. Prognostic value of changes in galectin-3 levels over time in patients with heart failure: data from CORONA and COACH. Circ Heart Fail. 2013;6:219–26.

    PubMed  Google Scholar 

  57. Yu L, Ruifrok WP, Meissner M, et al. Genetic and pharmacological inhibition of galectin-3 prevents cardiac remodeling by interfering with myocardial fibrogenesis. Circ Heart Fail. 2013;6:107–17. This article showed that galectin-3 could also serve as a “target for therapy”. Inhibition of galectin-3 resulted in attenuation of fibrosis in animal models of heart failure.

    CAS  PubMed  Google Scholar 

  58. Xu J, Kimball TR, Lorenz JN, et al. GDF15/MIC-1 functions as a protective and antihypertrophic factor released from the myocardium in association with SMAD protein activation. Circ Res. 2006;98:342–50. The authors identified GDF-15 as a novel antagonist of hypertrophic response.

    CAS  PubMed  Google Scholar 

  59. Kempf T, Eden M, Strelau J, et al. The transforming growth factor-beta superfamily member growth-differentiation factor-15 protects the heart from ischemia/reperfusion injury. Circ Res. 2006;98:351–60.

    CAS  PubMed  Google Scholar 

  60. de Jager SC, Bermudez B, Bot I, et al. Growth differentiation factor 15 deficiency protects against atherosclerosis by attenuating CCR2-mediated macrophage chemotaxis. J Exp Med. 2011;208:217–25.

    PubMed Central  PubMed  Google Scholar 

  61. Kempf T, von Haehling S, Peter T, et al. Prognostic utility of growth differentiation factor-15 in patients with chronic heart failure. J Am Coll Cardiol. 2007;50:1054–60.

    CAS  PubMed  Google Scholar 

  62. Foley PW, Stegemann B, Ng K, et al. Growth differentiation factor-15 predicts mortality and morbidity after cardiac resynchronization therapy. Eur Heart J. 2009;30:2749–57.

    CAS  PubMed Central  PubMed  Google Scholar 

  63. Anand IS, Kempf T, Rector TS, et al. Serial measurement of growth-differentiation factor-15 in heart failure: relation to disease severity and prognosis in the Valsartan Heart Failure Trial. Circulation. 2010;122:1387–95.

    CAS  PubMed  Google Scholar 

  64. Lok DJ, Klip IT, Lok SI, et al. Incremental prognostic power of novel biomarkers (growth-differentiation factor-15, high-sensitivity C-reactive protein, galectin-3, and high-sensitivity troponin-T) in patients with advanced chronic heart failure. Am J Cardiol. 2013;112:831–7.

    CAS  PubMed  Google Scholar 

  65. Izumiya Y, Hanatani S, Kimura Y, et al. Growth differentiation factor-15 is a useful prognostic marker in patients with heart failure with preserved ejection fraction. Can J Cardiol. 2014;30:338–44.

    PubMed  Google Scholar 

  66. Doust JA, Pietrzak E, Dobson A, Glasziou P. How well does B-type natriuretic peptide predict death and cardiac events in patients with heart failure: systematic review. BMJ. 2005;330:625. This systematic review concludes that BNP is a strong prognostic factor in patients with heart failure.

    CAS  PubMed Central  PubMed  Google Scholar 

  67. Brunner-La Rocca HP, Kaye DM, Woods RL, Hastings J, Esler MD. Effects of intravenous brain natriuretic peptide on regional sympathetic activity in patients with chronic heart failure as compared with healthy control subjects. J Am Coll Cardiol. 2001;37:1221–7.

    CAS  PubMed  Google Scholar 

  68. Kimura K, Yamaguchi Y, Horii M, et al. ANP is cleared much faster than BNP in patients with congestive heart failure. Eur J Clin Pharmacol. 2007;63:699–702.

    CAS  PubMed  Google Scholar 

  69. Moertl D, Berger R, Struck J, et al. Comparison of midregional pro-atrial and B-type natriuretic peptides in chronic heart failure: influencing factors, detection of left ventricular systolic dysfunction, and prediction of death. J Am Coll Cardiol. 2009;53:1783–90. MR-proANP was found to be a better predictor for death in patients with chronic heart failure, compared to BNP and NT-proBNP.

    CAS  PubMed  Google Scholar 

  70. Morgenthaler NG, Struck J, Thomas B, Bergmann A. Immunoluminometric assay for the midregion of pro-atrial natriuretic peptide in human plasma. Clin Chem. 2004;50:234–6.

    CAS  PubMed  Google Scholar 

  71. Mueller T, Gegenhuber A, Poelz W, Haltmayer M. Diagnostic accuracy of B type natriuretic peptide and amino terminal proBNP in the emergency diagnosis of heart failure. Heart. 2005;91:606–12.

    CAS  PubMed Central  PubMed  Google Scholar 

  72. Maisel A, Hollander JE, Guss D, et al. Primary results of the Rapid Emergency Department Heart Failure Outpatient Trial (REDHOT). A multicenter study of B-type natriuretic peptide levels, emergency department decision making, and outcomes in patients presenting with shortness of breath. J Am Coll Cardiol. 2004;44:1328–33.

    PubMed  Google Scholar 

  73. Baggish AL, van Kimmenade RR, Pinto Y, et al. New York Heart Association class versus amino-terminal pro-B type natriuretic peptide for acute heart failure prognosis. Biomarkers. 2010;15:307–14.

    CAS  PubMed  Google Scholar 

  74. Bayes-Genis A, Santalo-Bel M, Zapico-Muniz E, et al. N-terminal probrain natriuretic peptide (NT-proBNP) in the emergency diagnosis and in-hospital monitoring of patients with dyspnoea and ventricular dysfunction. Eur J Heart Fail. 2004;6:301–8.

    CAS  PubMed  Google Scholar 

  75. Bettencourt P, Azevedo A, Pimenta J, Frioes F, Ferreira S, Ferreira A. N-terminal-pro-brain natriuretic peptide predicts outcome after hospital discharge in heart failure patients. Circulation. 2004;110:2168–74.

    CAS  PubMed  Google Scholar 

  76. Januzzi Jr JL, Rehman SU, Mohammed AA, et al. Use of amino-terminal pro-B-type natriuretic peptide to guide outpatient therapy of patients with chronic left ventricular systolic dysfunction. J Am Coll Cardiol. 2011;58:1881–9.

    CAS  PubMed  Google Scholar 

  77. Schou M, Gustafsson F, Videbaek L, et al. Adding serial N-terminal pro brain natriuretic peptide measurements to optimal clinical management in outpatients with systolic heart failure: a multicentre randomized clinical trial (NorthStar monitoring study). Eur J Heart Fail. 2013;15:818–27. When serial measurements of NT-proBNP were added to optimal clinical management, this was not associated with improved outcome.

    CAS  PubMed  Google Scholar 

  78. Waldo SW, Beede J, Isakson S, et al. Pro-B-type natriuretic peptide levels in acute decompensated heart failure. J Am Coll Cardiol. 2008;51:1874–82.

    CAS  PubMed  Google Scholar 

  79. Mueller C, Laule-Kilian K, Scholer A, et al. B-type natriuretic peptide for acute dyspnea in patients with kidney disease: insights from a randomized comparison. Kidney Int. 2005;67:278–84.

    CAS  PubMed  Google Scholar 

  80. Das SR, Drazner MH, Dries DL, et al. Impact of body mass and body composition on circulating levels of natriuretic peptides: results from the Dallas Heart Study. Circulation. 2005;112:2163–8.

    CAS  PubMed  Google Scholar 

  81. O’Connor CM, Starling RC, Hernandez AF, et al. Effect of nesiritide in patients with acute decompensated heart failure. N Engl J Med. 2011;365:32–43. Infusion of nesiritide in a randomized controlled trial in 7,141 patients did not affect the rate of death or hospitalization.

    PubMed  Google Scholar 

  82. Seronde MF, Gayat E, Logeart D, et al. Comparison of the diagnostic and prognostic values of B-type and atrial-type natriuretic peptides in acute heart failure. Int J Cardiol. 2013;168:3404–11. This study of 710 patients evaluated the diagnostic and prognostic value of four natriuretic peptides and concluded that MR-proANP had the best prognostic value at five years.

    PubMed  Google Scholar 

  83. Testa G, Della-Morte D, Cacciatore F, et al. Precipitating factors in younger and older adults with decompensated chronic heart failure: are they different? J Am Geriatr Soc. 2013;61:1827–8.

    PubMed  Google Scholar 

  84. Steele IC, Nugent AM, Maguire S, et al. Cytokine profile in chronic cardiac failure. Eur J Clin Invest. 1996;26:1018–22.

    CAS  PubMed  Google Scholar 

  85. Pasceri V, Willerson JT, Yeh ET. Direct proinflammatory effect of C-reactive protein on human endothelial cells. Circulation. 2000;102:2165–8.

    CAS  PubMed  Google Scholar 

  86. Du Clos TW, Mold C. C-reactive protein: an activator of innate immunity and a modulator of adaptive immunity. Immunol Res. 2004;30:261–77.

    PubMed  Google Scholar 

  87. Zairis MN, Tsiaousis GZ, Georgilas AT, et al. Multimarker strategy for the prediction of 31 days cardiac death in patients with acutely decompensated chronic heart failure. Int J Cardiol. 2010;141:284–90.

    PubMed  Google Scholar 

  88. Kalogeropoulos AP, Tang WH, Hsu A, et al. High sensitivity C-reactive protein in acute heart failure: insights from the ASCEND-HF trial. J Card Fail. 2014;20:319–26.

  89. Lourenco P, Paulo Araujo J, Paulo C, et al. Higher C-reactive protein predicts worse prognosis in acute heart failure only in noninfected patients. Clin Cardiol. 2010;33:708–14. CRP has prognostic value in acute decompensated heart failure patients without infection but not in patients with infectious complications.

    PubMed  Google Scholar 

  90. Delevaux I, Andre M, Colombier M, et al. Can procalcitonin measurement help in differentiating between bacterial infection and other kinds of inflammatory processes? Ann Rheum Dis. 2003;62:337–40.

    CAS  PubMed Central  PubMed  Google Scholar 

  91. Picariello C, Lazzeri C, Valente S, Chiostri M, Gensini GF. Procalcitonin in acute cardiac patients. Intern Emerg Med. 2011;6:245–52.

    PubMed  Google Scholar 

  92. Picariello C, Lazzeri C, Valente S, Chiostri M, Attana P, Gensini GF. Kinetics of procalcitonin in cardiogenic shock and in septic shock. Preliminary data. Acute Card Care. 2010;12:96–101.

    PubMed  Google Scholar 

  93. Sinning CR, Sinning JM, Schulz A, et al. Association of serum procalcitonin with cardiovascular prognosis in coronary artery disease. Circ J. 2011;75:1184–91.

    PubMed  Google Scholar 

  94. Meisner M, Adina H, Schmidt J. Correlation of procalcitonin and C-reactive protein to inflammation, complications, and outcome during the intensive care unit course of multiple-trauma patients. Crit Care. 2006;10:R1.

    PubMed Central  PubMed  Google Scholar 

  95. Travaglino F, Russo V, De Berardinis B, et al. Thirty and ninety days mortality predictive value of admission and in-hospital procalcitonin and mid-regional pro-adrenomedullin testing in patients with dyspnea. Results from the VERyfing DYspnea trial. Am J Emerg Med. 2014;32:334–41. In patients with dyspnea, procalcitonin and MR-proADM improved risk stratification and management.

    PubMed  Google Scholar 

  96. Naffaa M, Makhoul BF, Tobia A, et al. Procalcitonin and interleukin 6 for predicting blood culture positivity in sepsis. Am J Emerg Med. 2014;32:448–51.

    PubMed  Google Scholar 

  97. Maisel A, Neath SX, Landsberg J, et al. Use of procalcitonin for the diagnosis of pneumonia in patients presenting with a chief complaint of dyspnoea: results from the BACH (Biomarkers in Acute Heart Failure) trial. Eur J Heart Fail. 2012;14:278–86.

    CAS  PubMed Central  PubMed  Google Scholar 

  98. Hausfater P, Garric S, Ayed SB, Rosenheim M, Bernard M, Riou B. Usefulness of procalcitonin as a marker of systemic infection in emergency department patients: a prospective study. Clin Infect Dis. 2002;34:895–901.

    CAS  PubMed  Google Scholar 

  99. Frangogiannis NG, Smith CW, Entman ML. The inflammatory response in myocardial infarction. Cardiovasc Res. 2002;53:31–47. This review describes the different components of the inflammatory response following myocardial infarction.

    CAS  PubMed  Google Scholar 

  100. Schuett H, Luchtefeld M, Grothusen C, Grote K, Schieffer B. How much is too much? Interleukin-6 and its signalling in atherosclerosis. Thromb Haemost. 2009;102:215–22.

    CAS  PubMed  Google Scholar 

  101. Rose-John S, Scheller J, Elson G, Jones SA. Interleukin-6 biology is coordinated by membrane-bound and soluble receptors: role in inflammation and cancer. J Leukoc Biol. 2006;80:227–36.

    CAS  PubMed  Google Scholar 

  102. Milo-Cotter O, Cotter-Davison B, Lombardi C, et al. Neurohormonal activation in acute heart failure: results from VERITAS. Cardiology. 2011;119:96–105.

    CAS  PubMed  Google Scholar 

  103. Lassus JP, Harjola VP, Peuhkurinen K, et al. Cystatin C, NT-proBNP, and inflammatory markers in acute heart failure: insights into the cardiorenal syndrome. Biomarkers. 2011;16:302–10.

    CAS  PubMed  Google Scholar 

  104. Miettinen KH, Lassus J, Harjola VP, et al. Prognostic role of pro- and anti-inflammatory cytokines and their polymorphisms in acute decompensated heart failure. Eur J Heart Fail. 2008;10:396–403. Proinflammatory and anti-inflammatory cytokines were measured in 423 patients with acute decompensated heart failure. IL-6, IL-10 and TNF-α provided important prognostic information in these patients.

    CAS  PubMed  Google Scholar 

  105. Pudil R, Tichy M, Andrys C, et al. Plasma interleukin-6 level is associated with NT-proBNP level and predicts short- and long-term mortality in patients with acute heart failure. Acta Med (Hradec Kralove). 2010;53:225–8.

    CAS  Google Scholar 

  106. Mann DL, Bristow MR. Mechanisms and models in heart failure: the biomechanical model and beyond. Circulation. 2005;111:2837–49. This review describes heart failure in different clinical model systems, like a cardiorenal model, a hemodynamic model, and a neurohormonal model.

    PubMed  Google Scholar 

  107. Agewall S, Giannitsis E, Jernberg T, Katus H. Troponin elevation in coronary vs. non-coronary disease. Eur Heart J. 2011;32:404–11.

    CAS  PubMed  Google Scholar 

  108. Adams 3rd JE, Bodor GS, Davila-Roman VG, et al. Cardiac troponin I. A marker with high specificity for cardiac injury. Circulation. 1993;88:101–6.

    PubMed  Google Scholar 

  109. Gaze DC, Collinson PO. Multiple molecular forms of circulating cardiac troponin: analytical and clinical significance. Ann Clin Biochem. 2008;45:349–55.

    CAS  PubMed  Google Scholar 

  110. Thygesen K, Alpert JS, White HD, et al. Universal definition of myocardial infarction. Circulation. 2007;116:2634–53.

    PubMed  Google Scholar 

  111. Steen H, Giannitsis E, Futterer S, Merten C, Juenger C, Katus HA. Cardiac troponin T at 96 hours after acute myocardial infarction correlates with infarct size and cardiac function. J Am Coll Cardiol. 2006;48:2192–4.

    CAS  PubMed  Google Scholar 

  112. Sato Y, Fujiwara H, Takatsu Y. Cardiac troponin and heart failure in the era of high-sensitivity assays. J Cardiol. 2012;60:160–7.

    PubMed  Google Scholar 

  113. Peacock 4th WF, De Marco T, Fonarow GC, et al. Cardiac troponin and outcome in acute heart failure. N Engl J Med. 2008;358:2117–26. Troponin levels were measured in 84,872 patients who were hospitalized for acute decompensated heart failure. A positive troponin test was associated with higher in-hospital mortality.

    CAS  PubMed  Google Scholar 

  114. de Antonio M, Lupon J, Galan A, et al. Head-to-head comparison of high-sensitivity troponin T and sensitive-contemporary troponin I regarding heart failure risk stratification. Clin Chim Acta. 2013;426:18–24. In a head-to-head comparison between troponin T and troponin I, troponin T showed better performance and was a better predictor for death.

    PubMed  Google Scholar 

  115. Kuwabara Y, Sato Y, Miyamoto T, et al. Persistently increased serum concentrations of cardiac troponin in patients with acutely decompensated heart failure are predictive of adverse outcomes. Circ J. 2007;71:1047–51.

    CAS  PubMed  Google Scholar 

  116. Damman K, Jaarsma T, Voors AA, et al. Both in- and out-hospital worsening of renal function predict outcome in patients with heart failure: results from the Coordinating Study Evaluating Outcome of Advising and Counseling in Heart Failure (COACH). Eur J Heart Fail. 2009;11:847–54.

    PubMed  Google Scholar 

  117. Damman K, Navis G, Voors AA, et al. Worsening renal function and prognosis in heart failure: systematic review and meta-analysis. J Card Fail. 2007;13:599–608. This systematic review and meta-analysis concludes that worsening renal function predicts substantially higher rates of mortality and hospitalization in patients with heart failure.

    PubMed  Google Scholar 

  118. Sarraf M, Masoumi A, Schrier RW. Cardiorenal syndrome in acute decompensated heart failure. Clin J Am Soc Nephrol. 2009;4:2013–26.

    CAS  PubMed  Google Scholar 

  119. Laterza OF, Price CP, Scott MG. Cystatin C: an improved estimator of glomerular filtration rate? Clin Chem. 2002;48:699–707.

    CAS  PubMed  Google Scholar 

  120. Fliser D, Ritz E. Serum cystatin C concentration as a marker of renal dysfunction in the elderly. Am J Kidney Dis. 2001;37:79–83.

    CAS  PubMed  Google Scholar 

  121. Manzano-Fernandez S, Boronat-Garcia M, Albaladejo-Oton MD, et al. Complementary prognostic value of cystatin C, N-terminal pro-B-type natriuretic Peptide and cardiac troponin T in patients with acute heart failure. Am J Cardiol. 2009;103:1753–9.

    CAS  PubMed  Google Scholar 

  122. Lassus J, Harjola VP, Sund R, et al. Prognostic value of cystatin C in acute heart failure in relation to other markers of renal function and NT-proBNP. Eur Heart J. 2007;28:1841–7.

    CAS  PubMed  Google Scholar 

  123. Maisel AS, Mueller C, Fitzgerald R, et al. Prognostic utility of plasma neutrophil gelatinase-associated lipocalin in patients with acute heart failure: the NGAL EvaLuation Along with B-type NaTriuretic Peptide in acutely decompensated heart failure (GALLANT) trial. Eur J Heart Fail. 2011;13:846–51. In this study, NGAL was measured in 186 patients with acute decompensated heart failure. Plasma NGAL was found to be a strong predictor for 30-days outcome and improved reclassification over BNP.

    CAS  PubMed Central  PubMed  Google Scholar 

  124. Aghel A, Shrestha K, Mullens W, Borowski A, Tang WH. Serum neutrophil gelatinase-associated lipocalin (NGAL) in predicting worsening renal function in acute decompensated heart failure. J Card Fail. 2010;16:49–54.

    CAS  PubMed Central  PubMed  Google Scholar 

  125. Lassus J, Gayat E, Mueller C, et al. Incremental value of biomarkers to clinical variables for mortality prediction in acutely decompensated heart failure: the Multinational Observational Cohort on Acute Heart Failure (MOCA) study. Int J Cardiol. 2013;168:2186–94. In the MOCA study, 5,306 patients with acute decompensated heart failure were included. Biomarkers including sST2, MR-proADM, natriuretic peptides and CRP added prognostic value to clinical risk factors for predicting mortality.

    PubMed  Google Scholar 

  126. Zairis MN, Tsiaousis GZ, Georgilas AT, et al. Multimarker strategy for the prediction of 31 days cardiac death in patients with acutely decompensated chronic heart failure. Int J Cardiol. 2010;141:284–90. This multimarker study concluded that an increasing number of elevated biomarkers increased the risk of cardiac death gradually.

    PubMed  Google Scholar 

  127. Potocki M, Breidthardt T, Mueller A, et al. Copeptin and risk stratification in patients with acute dyspnea. Crit Care. 2010;14:R213.

    PubMed Central  PubMed  Google Scholar 

  128. Boffa GM, Zaninotto M, Sartor R, et al. Interleukin-6 and tumor necrosis factor-alpha as biochemical markers of heart failure: a head-to-head clinical comparison with B-type natriuretic peptide. J Cardiovasc Med (Hagerstown). 2009;10:758–64.

    Google Scholar 

  129. Redfield MM, Rodeheffer RJ, Jacobsen SJ, Mahoney DW, Bailey KR, Burnett Jr JC. Plasma brain natriuretic peptide concentration: impact of age and gender. J Am Coll Cardiol. 2002;40:976–82.

    CAS  PubMed  Google Scholar 

  130. Hong M, Yan Q, Tao B, et al. Estradiol, progesterone and testosterone exposures affect the atrial natriuretic peptide gene expression in vivo in rats. Biol Chem Hoppe Seyler. 1992;373:213–8.

    CAS  PubMed  Google Scholar 

  131. Deng Y, Kaufman S. The influence of reproductive hormones on ANF release by rat atria. Life Sci. 1993;53:689–96.

    CAS  PubMed  Google Scholar 

  132. Ho JE, Gona P, Pencina MJ, et al. Discriminating clinical features of heart failure with preserved vs. reduced ejection fraction in the community. Eur Heart J. 2012;33:1734–41.

    PubMed Central  PubMed  Google Scholar 

  133. Meyer S, van der Meer P, van Deursen VM, et al. Neurohormonal and clinical sex differences in heart failure. Eur Heart J. 2013;34:2538–47. Female patients presenting with heart failure have a different clinical presentation and better outcome compared to male patients.

    CAS  PubMed  Google Scholar 

  134. de Boer RA, van Veldhuisen DJ, Gansevoort RT, et al. The fibrosis marker galectin-3 and outcome in the general population. J Intern Med. 2012;272:55–64. In this large community based cohort, galectin-3 was found to be associated with age and various risk factors of cardiovascular disease, with a strong gender interaction.

    PubMed  Google Scholar 

  135. Coglianese EE, Larson MG, Vasan RS, et al. Distribution and clinical correlates of the interleukin receptor family member soluble ST2 in the Framingham Heart Study. Clin Chem. 2012;58:1673–81.

    CAS  PubMed Central  PubMed  Google Scholar 

  136. Melander O, Maisel AS, Almgren P, et al. Plasma proneurotensin and incidence of diabetes, cardiovascular disease, breast cancer, and mortality. JAMA. 2012;308:1469–75.

    CAS  PubMed  Google Scholar 

  137. Montagnana M, Danese E, Giudici S, et al. HE4 in ovarian cancer: from discovery to clinical application. Adv Clin Chem. 2011;55:1–20.

    CAS  PubMed  Google Scholar 

  138. de Boer RA, Cao Q, Postmus D, et al. The WAP four-disulfide core domain protein HE4: a novel biomarker for heart failure. JACC Heart Fail. 2013;1:164–9. In the COACH trial, HE4 improved net reclassification when it was added to the clinical model and was correlated with markers of heart failure severity.

    PubMed  Google Scholar 

  139. LeBleu VS, Teng Y, O’Connell JT, et al. Identification of human epididymis protein-4 as a fibroblast-derived mediator of fibrosis. Nat Med. 2013;19:227–31. HE4 was found to be a new potential biomarker of (renal) fibrosis and could serve as a new therapeutic target.

    CAS  PubMed  Google Scholar 

  140. Smith GL, Lichtman JH, Bracken MB, et al. Renal impairment and outcomes in heart failure: systematic review and meta-analysis. J Am Coll Cardiol. 2006;47:1987–96.

    PubMed  Google Scholar 

  141. Del Ry S, Passino C, Maltinti M, Emdin M, Giannessi D. C-type natriuretic peptide plasma levels increase in patients with chronic heart failure as a function of clinical severity. Eur J Heart Fail. 2005;7:1145–8.

    PubMed  Google Scholar 

  142. Wu C, Wu F, Pan J, Morser J, Wu Q. Furin-mediated processing of Pro-C-type natriuretic peptide. J Biol Chem. 2003;278:25847–52.

    CAS  PubMed  Google Scholar 

  143. Zakeri R, Sangaralingham SJ, Sandberg SM, Heublein DM, Scott CG, Burnett Jr JC. Urinary C-type natriuretic peptide: a new heart failure biomarker. JACC Heart Fail. 2013;1:170–7. Urinary C-type natriuretic peptide was detected in patients with heart failure and may have clinical utility as a biomarker.

    PubMed  Google Scholar 

  144. Tromp J, van der Pol A, Klip IT, et al. The Fibrosis Marker Syndecan-1 and Outcome in Heart Failure Patients with Reduced and Preserved Ejection Fraction. Circ Heart Fail. 2014;7:457–62. Syndecan-1 levels correlate with biomarkers of fibrosis and was a predictor for outcome in patients with HFpEF but not in HFrEF.

  145. Castillo-Martinez L, Colin-Ramirez E, Orea-Tejeda A, et al. Cachexia assessed by bioimpedance vector analysis as a prognostic indicator in chronic stable heart failure patients. Nutrition. 2012;28:886–91.

    PubMed  Google Scholar 

  146. Di Somma S, Navarin S, Giordano S, et al. The emerging role of biomarkers and bio-impedance in evaluating hydration status in patients with acute heart failure. Clin Chem Lab Med. 2012;50:2093–105.

    PubMed  Google Scholar 

  147. Di Somma S, Lalle I, Magrini L, et al. Additive diagnostic and prognostic value of Bioelectrical Impedance Vector Analysis (BIVA) to brain natriuretic peptide ‘grey-zone’ in patients with acute heart failure in the emergency department. Eur Heart J Acute Cardiovasc Care. 2014;3:167–75. In 270 patients with acute heart failure, Bioelectrical Impedance Vector Analysis provided additive 30-day prognostic value to BNP.

  148. Morrow DA, de Lemos JA. Benchmarks for the assessment of novel cardiovascular biomarkers. Circulation. 2007;115:949–52.

    PubMed  Google Scholar 

  149. van Kimmenade RR, Januzzi Jr JL. Emerging biomarkers in heart failure. Clin Chem. 2012;58:127–38.

    PubMed  Google Scholar 

  150. Wang TJ, Gona P, Larson MG, et al. Multiple biomarkers for the prediction of first major cardiovascular events and death. N Engl J Med. 2006;355:2631–9.

    CAS  PubMed  Google Scholar 

  151. Cohen Freue GV, Meredith A, Smith D, et al. Computational biomarker pipeline from discovery to clinical implementation: plasma proteomic biomarkers for cardiac transplantation. PLoS Comput Biol. 2013;9:e1002963. Quantitive proteomics has led to discovery of new biomarkers for various diseases, thus, providing new treatments and diagnostics. This proposed computational pipeline is applicable to other biomarker proteomic studies.

    PubMed Central  PubMed  Google Scholar 

  152. Scrutinio D, Ammirati E, Guida P, et al. Clinical utility of N-terminal pro-B-type natriuretic peptide for risk stratification of patients with acute decompensated heart failure. Derivation and validation of the ADHF/NT-proBNP risk score. Int J Cardiol. 2013;168:2120–6.

    PubMed  Google Scholar 

  153. Maisel A, Xue Y, Shah K, et al. Increased 90-day mortality in patients with acute heart failure with elevated copeptin: secondary results from the Biomarkers in Acute Heart Failure (BACH) study. Circ Heart Fail. 2011;4:613–20.

    CAS  PubMed  Google Scholar 

Download references

Compliance with Ethics Guidelines

Conflict of Interest

A. Rogier van der Velde declares that he has no conflict of interest.

Wouter C. Meijers declares that he has no conflict of interest. The University Medical Center Groningen, where the authors are employed, has received research grants from BG Medicine.

Rudolf A. de Boer has received research grants from Abbott and Netherlands Organisation for Scientific Research, NWO VIDI grant (No. 917.13.350), consultancy and/or speaker fees from Abbott, AstraZeneca, Biomérieux, BG Medicine, Pfizer, Baxter, and Novartis, and has ownership interest in Pectacea.

Human and Animal Rights and Informed Consent

This article does not contain any studies with human or animal subjects performed by any of the authors.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Rudolf A. de Boer.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

van der Velde, A.R., Meijers, W.C. & de Boer, R.A. Biomarkers for Risk Prediction in Acute Decompensated Heart Failure. Curr Heart Fail Rep 11, 246–259 (2014). https://doi.org/10.1007/s11897-014-0207-7

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11897-014-0207-7

Keywords

Navigation